Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Apr 24.
Published in final edited form as: Breast Cancer Res Treat. 2008 May 8;114(3):559–568. doi: 10.1007/s10549-008-0028-z

DNA hypermethylation and clinicopathological features in breast cancer: the Western New York Exposures and Breast Cancer (WEB) Study

Meng Hua Tao 1,, Peter G Shields 2,, Jing Nie 3, Amy Millen 4, Christine B Ambrosone 5, Stephen B Edge 6, Shiva S Krishnan 7, Catalin Marian 8, Bin Xie 9, Janet Winston 10, Dominica Vito 11, Maurizio Trevisan 12, Jo L Freudenheim 13
PMCID: PMC4408917  NIHMSID: NIHMS677526  PMID: 18463976

Abstract

Aberrant DNA hypermethylation of gene promoter regions has been increasingly recognized as a common molecular alteration in carcinogenesis. We evaluated the association between major clinicopathological features and hypermethylation of genes in tumors among 803 incidence breast cancer cases from a large population-based case–control study conducted in Western New York State. DNA samples were isolated from archive paraffin embedded tumor tissue and were analyzed for hypermethylation status of the E-cadherin, p16, and RAR-β2 genes using real time methylation-specific polymerase chain reaction. The frequencies of hypermethylation were 20.0% for E-cadherin, 25.9% for p16, and 27.5% for RAR-β2 genes. For postmenopausal women, hypermethylation of E-cadherin tended to be more likely in progesterone receptor (PR) negative than in PR-positive tumors (odds ratio (OR), 1.41; 95% confidence interval (CI), 0.91–2.18). Hypermethylation of p16 tended to be more frequent among estrogen receptor (ER) negative cases than ER-positive cases (OR, 1.51; 95% CI, 1.01–2.32). Hypermethylation of RAR-β2 gene was inversely associated with histological and nuclear grade of breast cancer.

Keywords: Hypermethylation, Estrogen receptor, Progesterone receptor, Breast cancer, Epidemiology

Introduction

Aberrant DNA hypermethylation has been increasingly recognized as a frequent molecular alteration in cancer [1, 2]. This epigenetic modification occurs at the cytosines of CpG dinucleotide-rich regions, which are mostly unmethylated in normal tissues. Hypermethylation of CpG islands in gene promoter regions of many tumor suppressor and DNA repair genes is associated with chromatin condensation, delaying replication, inhibiting initiation of transcription and silencing of genes [3]. For breast tumors, there is evidence of hypermethylation of functionally important genes including those involved in DNA repair (BRCA1) [4], cell cycle regulation (p16) [3], cell adhesion (E-cadherin) [5], hormone and receptor-mediated cell signaling (ER (estrogen receptor) and RAR-β2 (retinoic acid-binding receptor-β2)) [6], regulation of cell transcription (HOXA5 (homeo box A5)) [7], and other functions [6]. There are suggestions that aberrant hypermethylation may be useful as a biomarker, with implications for breast cancer etiology, diagnosis and management.

Recent studies have focused on identifying the gene-specific hypermethylation profile of different tumors [3, 812]. Some studies have evaluated the association between gene hypermethylation and biological or clinical properties of breast tumors [1319]. An association between CpG island hypermethylation of p16 and RAR-β2 genes and poorly differentiated breast tumors was found in two previous studies [14, 19]. Promoter hypermethylation of PTEN (phosphatase and tensin homologue deleted on chromosome 10) was associated with larger tumor size and higher histologic grade [17]. Methylation of GSTP1 (glutathione S-transferase π 1) and/or RAR-β2 was found to be associated with breast cancer cases with sentinel lymph node metastasis [15]; and HIN-1 (high in normal-1) and RAR-β2 had greatly higher methylation frequencies in bone, brain, and lung metastases than the primary breast tumors [18]. Some studies reported no apparent association between methylation distribution phenotypes and tumor size, grade, stage, or lymph node status [13, 16]. Because of generally small sample sizes for existing studies, as well as differences in analytic methods and selection of genes, there still remains uncertainty regarding the relation between gene hypermethylation and breast tumor clinical characteristics.

The aim of the present study was to evaluate the association between the major clinicopathological features of breast cancer and methylation of three genes: p16, E-cadherin, and RAR-β2 among primary breast cancer cases from a large scale population-based case–control study. These genes are known to be important in breast cancer development and progression, and are frequently hypermethylated in breast tumors, leading to down-regulation of expression of their gene products.

Materials and methods

A population-based case–control study of breast cancer, the Western New York Exposures and Breast Cancer (WEB) Study was conducted in 1996–2001. Eligible cases were women diagnosed with primary, histologically confirmed, incident breast cancer, age 35–79, current residents of Erie or Niagara Counties in New York State, and with no previous cancer history other than nonmelanoma skin cancer. Among 1,627 eligible cases, 1,170 (72%) participated. Cases were interviewed within 1 year of diagnosis; most were interviewed within 3–6 months following diagnosis. All participants provided informed consent, and the study protocol was approved by the Institutional Review Boards of the University at Buffalo and of all the participating hospitals.

Extensive in-person interviews and self-administered questionnaires were used to ascertain information on potential confounding factors, breast cancer risk factors and anthropometric measures. Women were considered postmenopausal if their menses had ceased permanently and naturally, or if they had undergone any of the following conditions: a bilateral oophorectomy, a hysterectomy without removal of the ovaries and were older than 50, or radiation or other medical treatment which resulted in permanent cessation of their menses and were older than 55.

The pathological diagnosis of breast cancer was reconfirmed by a senior pathologist from Georgetown University. Information on cancer diagnosis, tumor size, histologic grade, and cancer stage (as measured by tumor-node-metastasis (TNM) stage) was abstracted from medical charts using a standard protocol. ER/PR status was determined by immunohistochemical analysis according to previous methods [20]. The 5 μm tissue were stained in DAKO Autostainer (DAKO, Carpentaria, CA) using the Dako Cytomation EnVision + system-HRP (DAB) kit. The Allred score was used to evaluate staining for ER and PR [20]. The staining signal was scored by estimating the proportion and average intensity of positive tumor cells. A proportion score (PS) ranging from 1 to 5 was assigned that represents the estimated proportion of positive tumor cells on the entire slide. An intensity score (IS) ranging from 1 to 3 was assigned that estimated the average staining intensity of positive tumor cells. The PS and IS were added to obtain a total score (TS) ranging from 2 to 8. A score of 3 or more was considered as positive, while a score of 2 or less was considered as negative. P53 mutation were identified using Affymetrix p53 Gene Chip System (Affymetrix, Santa Clara, CA) as previously described [21]. The presence of mutation was subsequently confirmed by bidirectional sequencing using the Big Dye Terminator v3.1 Cycle Sequencing Kit (Applied Biosystems, Foster City, CA) and the Mega BACE 1000 DNA Analysis System (GE Healthcare Bio-Sciences Corp., Piscataway, NJ) according to the manufacturers’ instructions.

Of the women with breast cancer in the WEB study, we was able to obtain archived tumor blocks for 922 of them. Of those we obtained, only tissue blocks with tumors were analyzed. Genomic tumor DNA isolated from tissue block was modified by bisulfite modification according to previous methods [22]. Briefly, genomic tumor DNA (2 μg in 20 μl DNA) was treated with of 3 M NaOH (2 μl) and incubated at 50°C for 20 min. Subsequently, 500 μl of the freshly prepared hydroquinone-bisulfite solution (2.5 M sodium metabisulfite, 2 M NaOH and 125 mM hydroquinone [pH 5.0]) was added to each DNA sample and placed on a 70°C heat block for 2 h in the dark. After the incubation, 1 ml of purification resin (Promega, Madison WI) was added to the DNA and subsequently processed through a minicolumn purification protocol (Promega, Madison WI) using a vacuum unit according to the manufacturer’s instructions.

Hypermethylation of E-cadherin, p16, and RAR-β2 was determined by real time methylation-specific polymerase chain reaction (MSP). As a control to check for modified viable DNA, we used a β-actin assay. If the β-actin result was negative, the DNA could not be used in subsequent assays, and re-modification was attempted. If β-actin was positive, then the other three genes were assayed immediately. We had hypermethylation results for 803 cases. The experiments were performed using the ABI 7900HT real time PCR system as previously described [23]. For each gene, fluorogenic PCR was carried out in a 10 μl reaction volume in a 384 well optical tray (AbGene, Surrey U.K.). The sequences of the primers and probe used to amplify and detect methylation were: 5′-CGATCGTATTCGGCGTTTGTTT-3′ (forward primer), 5′-CCGAAAAACTACGACTCCAAAAACC-3′ (reverse primer), and 5′-FAM-TCGTTCGGCGTTTTC-MGB-3′ (proble) for E-cadherin, 5′-GAGTTTTCGGTTGATTGGTTGGTT-3′ (forward primer), 5′-GCCGCACCTCCTCTAC-3′ (reverse primer), and 5′-FAM-CCCGAACCGCGACCGT-MGB-3′ (proble) for p16, and 5′-GAGTTGTTTGAGGATTGGGATGTC-3′ (forward primer), 5′-ACGATACCCAAACAAACCCTACTC-3′ (reverse primer), and 5′-FAM-ATCGCTCGCGTTCTC-MGB-3′ (proble) for RAR-β2. Each reaction contained 5 μl of Taqman Universal Master Mix (2×), 4.5 μl of bisulfite treated DNA and 0.5 μl of a 60× assay by design premix that was designed for each respective gene (Applied Biosystems, Carlsbad CA). Thermal cycling started with an initial 10 min denaturation at 95°C followed by cycling of 95°C for 15 s and 60°C for 1 min. This cycling was repeated 45 times and followed with a 5 min extension at 72°C whereupon the data was analyzed [24]. Each individual DNA sample was assayed in triplicate for each gene for quality control purposes. Additionally, as a positive control, universally methylated DNA (CpGenome; Norcross, GA) was used along with water blanks as a negative control.

The χ2 test was used to compare the distributions of the methylation status of individual genes and various features of breast cancer. Unconditional logistic regression was employed to estimate odds ratios (ORs) and 95% confidence intervals (CIs). We did a case–case comparison of those with to those without hypermethylation of a particular gene in their tumor, examining the likelihood of hypermethylation according to a tumor charateristic. All models were adjusted for age at diagnosis, education level, and race. Potential confounding effects from other demographic factors and known breast cancer risk factors, including age at menarche, age at menopause, parity, family history of breast cancer, and body mass index (BMI), were also examined and no appreciable confounding was observed. All statistical tests were based on two-sided probability. Statistical analyses were conducted using SAS Version 9.1 (SAS Institute, Cary, NC).

Results

Among women with available hypermethylation results, 736 (92.2%) cases were Caucasians and 67 (7.6%) were other racial groups including African-Americans (55), American Indian (3), Hispanic (3), Asian (2), and others (4). Approximately 70.5% of the cases were postmenopausal at diagnosis with mean age at diagnosis 57.0 years, and mean tumor size 1.8 cm. Additional primary clinical and pathologic characteristics of the women with breast cancer are shown in Table 1. The frequency of hypermethylation was 20.0% for E-cadherin, 25.9% for p16, and 27.5% for RAR-β2 gene (Table 2). Promoter region CpG hypermethylation for any one of the three genes was identified in 485 (60.4%) of 803 primary breast tumors. Fifty-one (6.4%) showed hypermethylation for two genes and only 1 (0.1%) tumor for all three genes. The frequencies of hypermethylation for E-cadherin, p16, and RAR-β2 genes were similar for pre- and postmenopausal cases.

Table 1.

Hypermethylation status of E-cadherin, p16, and RAR-β2 for pre- and postmenopausal women, WEB Study, 1996–2001a

Clinicopathologic factors Premenopausal wemen
Postmenopausal women
E-cadherin
p16
RAR-β2
E-cadherin
p16
RAR-β2
Yes No Yes No Yes No Yes No Yes No Yes No
Age at diagnosis (yr)
 < 50 45 164 49 160 58 151 8 30 4 34 14 24
 50–59 5 23 12 16 6 22 31 150 48 133 50 131
 60–69 38 169 60 147 56 151
 ≥70 34 106 35 105 37 103
P-value 0.65 0.03 0.48 0.41 0.12 0.63
Education (yr)
 < 12 3 5 2 6 2 6 10 36 13 33 13 33
 12 18 49 13 54 21 46 46 187 61 172 68 165
 ≥12 29 133 46 116 41 121 55 232 73 214 76 211
P-value 0.16 0.37 0.64 0.92 0.92 0.79
Tumor size (cm)b
 < 1.0 (0.9) 5 28 11 22 9 24 29 84 25 88 39 74
 1.0–1.7 (0.9–1.4) 15 56 14 57 18 53 22 120 40 102 38 104
 1.8–2.6 (1.5–1.9) 19 36 11 44 16 39 22 69 27 64 22 69
 ≥2.7 (2.0) 7 48 15 40 14 41 28 138 42 124 46 120
P-value 0.03 0.38 0.96 0.11 0.59 0.37
Metastatic disease at presentation
 No 28 120 37 111 39 109 87 318 109 296 107 298
 Yes 19 62 21 60 23 58 19 107 27 99 39 87
P-value 0.42 0.88 0.74 0.11 0.22 0.32
Vascular/lymph invasion
 No 30 117 35 112 39 108 78 309 105 282 102 285
 Yes 18 64 22 60 23 58 21 110 31 100 39 92
P-value 0.78 0.61 0.66 0.30 0.43 0.45
Histological grade
 Well 3 16 0 19 8 11 13 50 19 44 20 43
 Moderate 15 51 19 47 20 46 36 138 46 128 40 134
 Poor 26 84 26 84 24 86 44 159 55 148 53 150
P-value 0.75 0.03 0.13 0.97 0.85 0.39
Nuclear grade
 I 5 25 9 21 9 21 24 91 29 86 33 82
 II 19 56 18 57 22 53 41 149 49 141 47 143
 III 25 89 27 87 27 87 40 155 52 143 50 145
P-value 0.62 0.76 0.62 0.97 0.96 0.74
TNM stage
 0 7 22 7 22 11 18 10 52 14 48 17 45
 I 15 65 21 59 20 60 53 202 72 183 67 188
 IIa/IIb 21 67 17 71 20 68 26 128 40 114 48 106
 III/IV 2 21 9 14 10 13 4 20 2 22 8 16
P-value 0.40 0.25 0.12 0.71 0.18 0.69
ER status
 + 28 112 36 104 43 97 81 310 92 299 111 280
 − 16 64 22 58 16 64 29 124 48 105 39 114
P-value 1.0 0.77 0.08 0.64 0.06 0.50
PR status
 + 28 120 40 108 39 109 59 266 78 247 94 231
 − 16 51 15 52 19 48 46 156 56 146 53 149
P-value 0.40 0.47 0.76 0.20 0.34 0.50
ER/PR
 Both + 23 98 29 92 36 85 56 240 70 226 86 210
 Either + 10 32 17 25 8 34 22 85 27 80 29 78
 Both − 11 39 9 41 13 37 26 94 37 83 30 90
P-value 0.77 0.04 0.40 0.80 0.31 0.70
p53 mutation
 Wild 30 119 38 111 40 109 66 276 96 246 92 250
 Mutant 15 49 16 48 17 47 25 104 34 95 38 91
P-value 0.59 0.94 0.97 0.98 0.71 0.58
a

Subjects with missing values were excluded from the analysis

b

Cut-off point of tumor size among postmenopausal women

Table 2.

Frequencies (%) of hypermethylated genes, WEB Study, 1996–2001

Genes Overall Methylated cases (%) Premenopausal Methylated cases (%) Postmenopausal Methylated cases (%)
E-cadherin 161 (20.0) 50 (21.1) 111 (19.6)
p16 208 (25.9) 61 (25.7) 147 (26.0)
RAR-β2 221 (27.5) 64 (27.0) 157 (27.7)
None 266 (33.1) 77 (32.5) 189 (33.4)
Any one 485 (60.4) 146 (61.6) 339 (59.9)
Any two 51 (6.4) 13 (5.5) 38 (6.7)
Three 1 (0.1) 1 (0.4) 0 (0.0)

The relationship between individual gene hypermethylation status and clinical and pathologic features of breast cancer were evaluated stratifying by menopausal status in Table 3. Associations varied somewhat by menopausal status, but tests for multiplicative interaction were not significant (P > 0.05). For premenopausal women, while there was no association of hypermethylation with tumors stratified on ER status or PR status, in examination of tumors with either ER− or PR-positive status there was a higher likelihood of hypermethylation of p16 (OR, 2.21; 95% CI, 1.05–4.61), in a model adjusting for age at diagnosis, race, and education level. Among postmenopausal women, hypermethylation of E-cadherin tended to be more frequent among PR-negative cases (OR, 1.41, 95% CI, 0.91–2.18), although confidence intervals included the null. Patients with ER-negative tumors were more likely to have hypermethylation of the p16 gene (OR, 1.51, 95% CI, 1.01–2.32). No associations were observed for tumors that were both ER-negative and PR-negative and the hypermethylation of any gene. Histological grade was inversely associated with hypermethylation of RAR-β2 gene (OR, 0.59 and 0.69; 95% CI, 0.37–0.94 and 0.44–1.07, for moderate and poorly differentiated tumors, respectively); further tumors with poorer nuclear grade were inversely associated with hypermethylation of RAR-β2 gene. We observed no associations between hypermethylation of E-cadherin, p16, and RAR-β2 with tumor size, stage, metastases, or p53 mutation among both pre- and postmenopausal women.

Table 3.

Association of E-cadherin, p16, and RAR-β2 hypermethylation with selected clinicopathological factors for breast cancer, WEB Study, 1996–2001a

E-cadherin
ORb (95% CI) p16
ORb (95% CI) RAR-β2
ORb (95% CI)
Yes No Yes No Yes No
Pre-menopausal
Tumor size (cm)
 1.0 5 28 1.0 11 22 1.0 9 24 1.0
 1.0–1.7 15 56 1.43 (0.57–3.62) 14 57 0.37 (0.16–0.84) 18 53 0.97 (0.43–2.15)
 1.8–2.6 19 36 2.65 (1.06–6.62) 11 44 0.41 (0.17–0.97) 16 39 1.06 (0.46–2.45)
 ≥2.7 7 48 0.76 (0.26–2.23) 15 40 0.59 (0.26–1.34) 14 41 0.94 (0.40–2.19)
Metastatic disease at presentation
 No 28 120 1.0 37 111 1.0 39 109 1.0
 Yes 19 62 1.35 (0.69–2.63) 21 60 1.02 (0.55–1.91) 23 58 1.20 (0.65–2.23)
Vascular/lymph invasion
 No 30 117 1.0 35 112 1.0 39 108 1.0
 Yes 18 64 1.12 (0.57–2.18) 22 60 1.17 (0.63–2.18) 23 58 1.19 (0.64–2.18)
Histological grade
 Well 3 16 1.0 0 19 8 11 1.0
 Moderate 15 51 1.61 (0.64–4.04) 19 47 20 46 0.89 (0.42–1.91)
 Poor 26 84 1.74 (0.75–4.06) 26 84 24 86 0.61 (0.30–1.25)
Nuclear grade
 I 5 25 1.0 9 21 1.0 9 21 1.0
 II 19 56 2.28 (0.83–6.24) 18 57 0.64 (0.29–1.44) 22 53 0.88 (0.40–1.95)
 III 25 89 1.78 (0.67–4.73) 27 87 0.65 (0.31–1.38) 27 87 0.67 (0.31–1.45)
TNM stage
 0 7 22 1.0 7 22 1.0 11 18 1.0
 I 15 65 0.67 (0.28–1.62) 21 59 0.77 (0.34–1.74) 20 60 0.81 (0.36–1.84)
 IIa/IIb 21 67 0.94 (0.41–2.17) 17 71 0.52 (0.23–1.19) 20 68 0.70 (0.31–1.58)
 III/IV 2 21 0.25 (0.05–1.26) 9 14 1.52 (0.52–4.39) 10 13 2.03 (0.70–5.91)
ER status
 + 28 112 1.0 36 104 1.0 43 97 1.0
 − 16 64 1.06 (0.52–2.13) 22 58 1.07 (0.57–2.02) 16 64 0.62 (0.32–1.21)
PR status
 + 28 120 1.0 40 108 1.0 39 109 1.0
 − 16 51 1.42 (0.69–2.90) 15 52 0.74 (0.37–1.50) 19 48 1.26 (0.65–2.44)
ER/PR
 Both + 23 98 1.0 29 92 1.0 36 85 1.0
 Either + 10 32 1.34 (0.58–3.09) 17 25 2.21 (1.05–4.61) 8 34 0.54 (0.23–1.28)
 Both − 11 39 1.08 (0.48–2.44) 9 41 0.64 (0.28–1.49) 13 37 1.00 (0.47–2.12)
p53 mutation
 Wild 30 119 1.0 38 111 1.0 40 109 1.0
 Mutant 15 49 1.15 (0.56–2.34) 16 48 1.0 (0.50–1.98) 17 47 1.02 (0.52–2.00)
Postmenopausal
Tumor size (cm)
 < 0.9 29 84 1.0 25 88 1.0 39 74 1.0
 0.9–1.4 22 120 0.59 (0.33–1.06) 40 102 1.32 (0.79–2.21) 38 104 0.83 (0.51–1.37)
 1.5–1.9 22 69 1.04 (0.57–1.90) 27 64 1.43 (0.81–2.56) 22 69 0.73 (0.41–1.30)
 ≥2.0 28 138 0.67 (0.39–1.16) 42 124 1.17 (0.70–1.94) 46 120 0.88 (0.55–1.41)
Metastatic disease at presentation
 No 87 318 1.0 109 296 1.0 107 298 1.0
 Yes 19 107 0.67 (0.39–1.15) 27 99 0.75 (0.46–1.21) 39 87 1.24 (0.80–1.93)
Vascular/lymph invasion
 No 78 309 1.0 105 282 1.0 102 285 1.0
 Yes 21 110 0.77 (0.45–1.31) 31 100 0.84 (0.53–1.33) 39 92 1.18 (0.76–1.83)
Histological grade
 Well 13 50 1.0 19 44 1.0 20 43 1.0
 Moderate 36 138 1.32 (0.77–2.25) 46 128 1.12 (0.70–1.80) 40 134 0.59 (0.37–0.94)
 Poor 44 159 1.43 (0.86–2.40) 55 148 1.18 (0.74–1.86) 53 150 0.69 (0.44–1.07)
Nuclear grade
 I 24 91 1.0 29 86 1.0 33 82 1.0
 II 41 149 1.38 (0.82–2.34) 49 141 1.03 (0.64–1.64) 47 143 0.67 (0.42–1.05)
 III 40 155 1.38 (0.81–2.35) 52 143 1.11 (0.69–1.77) 50 145 0.68 (0.43–1.07)
TNM stage
 0 10 52 1.0 14 48 1.0 17 45 1.0
 I 53 202 0.99 (0.59–1.66) 72 183 1.19 (0.73–1.92) 67 188 1.02 (0.63–1.65)
 IIa/IIb 26 128 0.80 (0.44–1.45) 40 114 1.09 (0.63–1.86) 48 106 1.30 (0.77–2.19)
 III/IV 4 20 0.79 (0.25–2.50) 2 22 0.28 (0.06–1.25) 8 16 1.42 (0.56–3.62)
ER status
 + 81 310 1.0 92 299 1.0 111 280 1.0
 − 29 124 0.92 (0.57–1.48) 48 105 1.51 (1.01–2.32) 39 114 0.86 (0.56–1.32)
PR status
 + 59 266 1.0 78 247 1.0 94 231 1.0
 − 46 156 1.41 (0.91–2.18) 56 146 1.25 (0.83–1.87) 53 149 0.86 (0.58–1.28)
ER/PR
 Both + 56 240 1.0 70 226 1.0 86 210 1.0
 Either + 22 85 1.18 (0.68–2.04) 27 80 1.06 (0.64–1.76) 29 78 0.91 (0.56–1.48)
 Both − 26 94 1.27 (0.76–2.14) 37 83 1.42 (0.89–2.25) 30 90 0.81 (0.50–1.30)
p53 mutation
 Wild 66 276 1.0 96 246 1.0 92 250 1.0
 Mutant 25 104 1.01 (0.60–1.69) 34 95 0.91 (0.58–1.45) 38 91 1.13 (0.72–1.76)
a

Numbers for some analyses are less than total for entire group because of missing variables

b

Odds ratios (ORs) and 95% confidence intervals (CI) were estimated with unconditional logistic model adjusted for age at diagnosis for breast cancer, race, and education

We also evaluated hypermethylation in the three genes together (Table 4). There was a tendency for tumors with hypermethylation in at least one gene to be more likely to be PR-negative among postmenopausal women (OR, 1.39; 95% CI, 0.95–2.04), although the confidence interval include the null. Among premenopausal women, nodal invasion or metastatsis was associated with hypermethylation in at least one gene. No associations were observed between other clinicopathologic features of breast cancer and hypermethylation in at least one gene.

Table 4.

Association between clinicopathological factors and hypermethylation of at least one gene for breast cancer, WEB Study, 1996–2001a

All subjects
Premenopausal
Postmenopausal
Yes No ORb (95% CI) Yes No ORb (95% CI) Yes No ORb (95% CI)
Tumor size (cm)
 < 1.0 115 50 1.0 22 11 1.0 93 39 1.0
 1.0–1.4 110 58 0.87 (0.57–1.33) 28 17 0.63 (0.27–1.47) 82 41 0.97 (0.60–1.58)
 1.5–2.1 134 64 0.98 (0.65–1.47) 38 19 0.75 (0.33–1.70) 96 45 1.06 (0.66–1.69)
 ≥2.2 127 68 0.87 (0.58–1.31) 53 26 0.76 (0.36–1.63) 74 42 0.88 (0.54–1.45)
Metastatic disease at presentation
 No 367 186 1.0 93 55 1.0 274 131 1.0
 Yes 139 68 1.05 (0.74–1.48) 61 20 1.89 (1.02–3.50) 78 48 0.78 (0.51–1.19)
Vascular/lymph invasion
 No 349 185 1.0 92 55 1.0 257 130 1.0
 Yes 146 67 1.16 (0.82–1.63) 62 20 1.89 (1.03–3.47) 84 47 0.90 (0.60–1.37)
Histological grade
 Well 57 25 1.0 11 8 1.0 46 17 1.0
 Moderate 157 83 0.94 (0.64–1.36) 47 19 1.11 (0.52–2.39) 110 64 0.87 (0.57–1.35)
 Poor 212 101 1.05 (0.73–1.51) 71 39 0.84 (0.42–1.65) 141 62 1.17 (0.76–1.80)
Nuclear grade
 I 99 46 1.0 21 9 1.0 78 37 1.0
 II 173 92 0.84 (0.57–1.23) 51 24 0.85 (0.39–1.89) 122 68 0.82 (0.53–1.27)
 III 205 104 0.88 (0.61–1.29) 75 39 0.77 (0.37–1.62) 130 65 0.93 (0.60–1.45)
TNM stage
 0 57 34 1.0 22 7 1.0 35 27 1.0
 I 223 112 0.94 (0.64–1.39) 49 31 0.51 (0.22–1.15) 174 81 1.15 (0.74–1.79)
 IIa/IIb 160 82 0.95 (0.63–1.44) 57 31 0.59 (0.26–1.33) 103 51 1.13 (0.69–1.85)
 III/IV 33 14 1.13 (0.56–2.30) 19 4 1.55 (0.43–5.59) 14 10 0.77 (0.31–1.86)
ER status
 + 352 179 1.0 95 45 1.0 257 134 1.0
 − 158 75 1.11 (0.80–1.56) 52 28 0.95 (0.52–1.73) 106 47 1.22 (0.81–1.83)
PR status
 + 307 166 1.0 98 50 1.0 209 116 1.0
 − 189 80 1.31 (0.95–1.82) 46 21 1.20 (0.63–2.28) 143 59 1.39 (0.95–2.04)
ER/PR
 Both − 270 147 1.0 80 41 1.0 190 106 1.0
 Either + 106 43 1.35 (0.90–2.01) 31 11 1.42 (0.65–3.10) 75 32 1.34 (0.84–2.16)
 Both + 116 54 1.18 (0.81–1.72) 32 18 0.91 (0.46–1.83) 84 36 1.34 (0.85–2.11)
p53 mutation
 Wild 165 326 1.0 53 96 1.0 112 230 1.0
 Mutant 61 132 1.10 (0.77–1.57) 18 46 1.42 (0.74–2.71) 43 86 0.97 (0.63–1.49)
a

Numbers for some analyses are less than total for entire group because of missing variables

b

Odds ratios (ORs) and 95% confidence intervals (CI) were estimated with unconditional logistic model adjusted for age at diagnosis for breast cancer, race, and education

We analyzed the data among Caucasians alone; and the estimates and the confidence intervals were similar to results in the overall population (data not shown).

Discussion

To better understand the role of promoter hypermethylation status in the natural history of breast carcinogenesis and as a molecular predictor of disease progression, we evaluated the association between gene hypermethylation and clinicopathological characteristics of primary breast tumors. Promoter methylation for at least one gene was found in 66.9% of the breast tumors. Hypermethylation frequencies of individual genes reported in previous studies on breast cancer have varied widely. Frequencies for p16 and RAR-β2 genes in our sample were similar to those reported previously [14]. However, the frequency for E-cadherin hypermethylation (20.0%) was lower than other reports; in those the frequency of E-cadherin hypermethylation ranged between 39 and 80% [1416]. This variation may depend on the sensitivity of the MSP assay, differences in MSP assay design, and by sample size or other differences in the populations under study. In our study, we used the same assay conditions for each tumor DNA sample and positive and negative internal controls; our MSP analysis was reliable.

Encoded by the E-cadherin gene, the transmembrane glycoprotein E-cadherin is involved in maintaining homotypic cell-to-cell adhesion of differentiated epithelial tissues. Loss of E-cadherin expression has been related to loss of differentiation, increased invasiveness, and decreased patient survival [25, 26]. Although mutations and deletions in E-cadherin gene have been reported in cancers including lobular breast carcinoma [2729], in most breast carcinomas, E-cadherin mutations have been found to be rare or absent. Promoter hypermethylation of the gene might play a role in alteration of E-cadherin expression. Hypermethylation of the E-cadherin promoter has been shown to be associated with loss of E-cadherin expression in breast cancer cell lines and primary ductal and lobular breast cancers [25, 3032]. Previous studies have found a correlation between reduced E-cadherin expression and loss of ER and PR [33, 34]. Consistent with those investigations, a positive association between hypermethylation of E-cadherin gene and negative PR status was observed in our population. We did not see any association of E-cadherin hypermethylation with stage or metastasis.

The p16 gene, one of the most commonly inactivated tumor suppressor genes in human cancer [35], is a cyclin-dependent kinase inhibitor that regulates progression through the G1 phase of the cell-cycle [35]. Down-regulation of p16 expression caused by promoter hypermethylation occurs frequently in breast cancer. There is evidence that p16 hypermethylation is an early and likely critical step in breast cancer development [36, 37]. We found some tendency for hypermethylation in p16 to occur more often in ER-negative cancer patients than ER-positive among postmenopausal women. We did not observe associations of p16 hypermethylation with other clinic or pathological features of breast cancer.

The protein coded by RAR-β2 functions in inhibition of proliferation, apoptosis, and senescence. The gene is methylated frequently in breast cancer [14, 15, 18] and even normal breast tissue [38], which may result in loss of expression and a loss of control of cellular proliferation. Unlike previous studies, in which there was found a positive association between RAR-β2 hypermethylation and metastasis in breast tumor [15, 18], we found no associations between hypermethylation of RAR-β2 gene and metastasis and lymphovascular invasion. However, we found inverse associations between histological and nuclear grade and RAR-β2 hypermethylation among postmenopausal women.

The strengths of this study include the population-based study design and a relatively large sample size, leading to relatively stable risk estimates. Nevertheless, the statistical power in subgroups of our study remained limited due to the low frequencies of the hypermethylation, which limited our ability to identify weak associations. We cannot rule out the possibility that hypermethylation of genes other than those included in our study may be related to clinicopathological features of breast cancer. Further our inability to obtain the paraffin embedded breast tumor tissue for 24.7% of cases may have led to selection bias. Comparing with cases without breast tumor tissue, cases with breast tumor tissue had slightly younger age at diagnosis and higher TNM stage of breast tumor. However, they had similar tumor size, frequencies of histological grade, nuclear grade, ER and PR status.

In summary, our data suggest that E-cadherin hypermethylation is associated with higher histological grade and PR-negative tumors, p16 hypermethylation is associated with ER-negative tumors, and RAR-β2 hypermethylation is inversely related to histological and nuclear grade. Although there appears to be little support from results for a distinctive promoter hypermethylation phenotype in breast cancer, further research is needed to assess the association of these characteristics of tumors with other breast cancer risk factors to better understand their etiology.

Acknowledgments

This study would not have been possible without the support of all of the study participants and research staff of the Western New York Exposures and Breast Cancer (WEB) Study. Funding This work was funded by United States Public Health Service (USPHS) Grant Number R01CA92585 from the National Cancer Institute, and #DAMD 17030446 and #DAMD 179616202 from the DOD.

Contributor Information

Meng Hua Tao, Email: mtao@buffalo.edu, Department of Social and Preventive Medicine, School of Public Health and Health Professions, University at Buffalo, 270 Farber Hall, Buffalo, NY 14214, USA.

Peter G. Shields, Email: pgs2@georgetown.edu, Lombardi Cancer Center, Georgetown University Medical Center, 3800 Reservoir RD. NW, LL (s) Level, Room 150, Box 571465, Washington, DC 20057, USA

Jing Nie, Department of Social and Preventive Medicine, School of Public Health and Health Professions, University at Buffalo, 270 Farber Hall, Buffalo, NY 14214, USA.

Amy Millen, Department of Social and Preventive Medicine, School of Public Health and Health Professions, University at Buffalo, 270 Farber Hall, Buffalo, NY 14214, USA.

Christine B. Ambrosone, Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY 14263, USA

Stephen B. Edge, Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY 14263, USA

Shiva S. Krishnan, Lombardi Cancer Center, Georgetown University Medical Center, 3800 Reservoir RD. NW, LL (s) Level, Room 150, Box 571465, Washington, DC 20057, USA

Catalin Marian, Lombardi Cancer Center, Georgetown University Medical Center, 3800 Reservoir RD. NW, LL (s) Level, Room 150, Box 571465, Washington, DC 20057, USA.

Bin Xie, Lombardi Cancer Center, Georgetown University Medical Center, 3800 Reservoir RD. NW, LL (s) Level, Room 150, Box 571465, Washington, DC 20057, USA.

Janet Winston, Potomac Hospital, Woodbridge, VA 22191, USA.

Dominica Vito, Department of Social and Preventive Medicine, School of Public Health and Health Professions, University at Buffalo, 270 Farber Hall, Buffalo, NY 14214, USA.

Maurizio Trevisan, Department of Social and Preventive Medicine, School of Public Health and Health Professions, University at Buffalo, 270 Farber Hall, Buffalo, NY 14214, USA.

Jo L. Freudenheim, Department of Social and Preventive Medicine, School of Public Health and Health Professions, University at Buffalo, 270 Farber Hall, Buffalo, NY 14214, USA

References

  • 1.Wajed SA, Laird PW, DeMeester TR. DNA methylation: an alternative pathway to cancer. Ann Surg. 2001;234:10–20. doi: 10.1097/00000658-200107000-00003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Baylin SB, Herman JG. DNA hypermethylation in tumori-genesis: epigenetics joins genetics. Trends Genet. 2000;16:168–174. doi: 10.1016/s0168-9525(99)01971-x. [DOI] [PubMed] [Google Scholar]
  • 3.Esteller M, Corn PG, Baylin SB, Herman JG. A gene hypermethylation profile of human cancer. Cancer Res. 2001;61:3225–3229. [PubMed] [Google Scholar]
  • 4.Esteller M, Silva JM, Dominguez G, et al. Promoter hypermethylation and BRCA1 inactivation in sporadic breast and ovarian tumors. J Natl Cancer Inst. 2000;92:564–569. doi: 10.1093/jnci/92.7.564. [DOI] [PubMed] [Google Scholar]
  • 5.Graff JR, Herman JG, Lapidus RG, et al. E-cadherin expression is silenced by DNA hypermethylation in human breast and prostate carcinomas. Cancer Res. 1995;55:5195–5199. [PubMed] [Google Scholar]
  • 6.Widschwendter M, Peter AJ. DNA methylation and breast carcinogenesis. Oncogene. 2002;21:5462–5482. doi: 10.1038/sj.onc.1205606. [DOI] [PubMed] [Google Scholar]
  • 7.Raman V, Martensen SA, Reisman D, et al. Compromised HOXA5 function can limit p53 expression in human breast tumors. Nature. 2000;405:974–978. doi: 10.1038/35016125. [DOI] [PubMed] [Google Scholar]
  • 8.Wiley A, Katasaros D, Chen H, et al. Aberrant promoter methylation of multiple genes in malignant ovarian tumors and in ovarian tumors with low malignant potential. Cancer. 2006;107:299–308. doi: 10.1002/cncr.21992. [DOI] [PubMed] [Google Scholar]
  • 9.Toyota M, Ahuja N, Suzuki H, et al. Aberrant methylation in gastric cancer associated with the CpG island methylator phenotype. Cancer Res. 1999;59:5438–5442. [PubMed] [Google Scholar]
  • 10.Bannon K, Yanokura M, Susumu N, et al. Relationship of the aberrant DNA hypermethylation of cancer-related genes with carcinogenesis of endometrial cancer. Oncol Rep. 2006;16:1189–1196. [PubMed] [Google Scholar]
  • 11.Jeong DH, Youm MY, Kim YN, et al. Promoter methylation of p16, DAPK, CDH1, and TIMP-3 genes in cervical cancer: correlation with clinicopathologic characteristics. Int J Gynecol Cancer. 2006;16:1234–1240. doi: 10.1111/j.1525-1438.2006.00522.x. [DOI] [PubMed] [Google Scholar]
  • 12.Belinsky SA, Liechty KC, Gentry FD, et al. Promoter hypermthylation of multiple genes in sputum precedes lung cancer incidence in a high-risk cohort. Cancer Res. 2006;66:3338–3344. doi: 10.1158/0008-5472.CAN-05-3408. [DOI] [PubMed] [Google Scholar]
  • 13.Bae YK, Brown A, Garrett E, et al. Hypermethylation in histologically distinct classes of breast cancer. Clin Cancer Res. 2004;10:5988–6005. doi: 10.1158/1078-0432.CCR-04-0667. [DOI] [PubMed] [Google Scholar]
  • 14.Li SY, Rong M, Iacopetta B. DNA hypermethylation in breast cancer and its association with clinicopathological features. Cancer Lett. 2006;237:272–280. doi: 10.1016/j.canlet.2005.06.011. [DOI] [PubMed] [Google Scholar]
  • 15.Shinozaki M, Hoon DSB, Giuliano AE, et al. Distinct hypermethylation profile of primary breast cancer is associated with sentinel lymph node metastasis. Clin Cancer Res. 2005;11:2156–2162. doi: 10.1158/1078-0432.CCR-04-1810. [DOI] [PubMed] [Google Scholar]
  • 16.Parrella P, Poeta ML, Gallo AP, et al. Nonrandom distribution of aberrant promoter methylation of cancer-related genes in sporadic breast tumors. Clin Cancer Res. 2004;10:5349–5354. doi: 10.1158/1078-0432.CCR-04-0555. [DOI] [PubMed] [Google Scholar]
  • 17.Garcia JM, Silva J, Pena C, et al. Promoter methylation of the PTEN gene is a common molecular change in breast cancer. Genes Chromosomes Cancer. 2004;41:117–124. doi: 10.1002/gcc.20062. [DOI] [PubMed] [Google Scholar]
  • 18.Mehrotra J, Vali M, McVeigh M, et al. Very high frequency of hypermthylated genes in breast cancer metastasis to the bone, brain, and lung. Clin Cancer Res. 2004;10:3104–3109. doi: 10.1158/1078-0432.ccr-03-0118. [DOI] [PubMed] [Google Scholar]
  • 19.Yan PS, Perry MR, Laux DE, Asare AL, Caldwell CW, Huang TH. CpG island arrays: an application toward deciphering epigenetic signatures of breast cancer. Clin Cancer Res. 2006;6:1432–1438. [PubMed] [Google Scholar]
  • 20.Allred DC, Harvey JM, Berardo M, Clark GM. Prognostic and predictive factors in breast cancer by immumohistochemical analysis. Mod Pathol. 1998;11:155–168. [PubMed] [Google Scholar]
  • 21.Tennis M, Krishnan S, Bonner M, et al. p53 Mutation analysis in breast tumors by a DNA microarray method. Cancer Epidemiol Biomarkers Prev. 2006;15:80–85. doi: 10.1158/1055-9965.EPI-05-0444. [DOI] [PubMed] [Google Scholar]
  • 22.Jeronimo C, Usadel H, Henrique R, et al. Quantitation of GSTP1 methylation in non-neoplastic prostatic tissue and organ-confined prostate adenocarcinoma. J Natl Cancer Inst. 2001;93:1447–1452. doi: 10.1093/jnci/93.22.1747. [DOI] [PubMed] [Google Scholar]
  • 23.Harden SV, Tokumaru Y, Westra WH, et al. Gene promoter hypermethylation in tumors and lymph nodes of stage 1 lung cancer patients. Clin Cancer Res. 2003;9:1370–1375. [PubMed] [Google Scholar]
  • 24.Usadel H, Brabender J, Danenberg KD, et al. Quantitative adenomatous polyposis coli promoter methylation analysis in tumor tissue, serum, and plasma DNA of patients with lung cancer. Cancer Res. 2002;62:371–375. [PubMed] [Google Scholar]
  • 25.Graff JR, Gabrelson E, Fujii H, Baylin SB, Herman JG. Methylation patterns of the E-cadherin 5′ CpG island are unstable and reflect the dynamic heterogeneous loss of E-cadherin expression during metastatic progression. J Biol Chem. 2000;275:2727–2732. doi: 10.1074/jbc.275.4.2727. [DOI] [PubMed] [Google Scholar]
  • 26.Hazan NM, Qiao R, Keren R, Badano I, Suyama K. Cadherin switch in tumor progression. Ann N Y Acad Sci. 2004;1014:155–163. doi: 10.1196/annals.1294.016. [DOI] [PubMed] [Google Scholar]
  • 27.Rieger-Christ KM, Pezza JA, Dugan JM, et al. Disparate E-cadherin mutations in LCIS and associated invasive breast carcinomas. Mol Pathol. 2001;54:91–97. doi: 10.1136/mp.54.2.91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.More H, Humar B, Weber W, et al. Identification of seven novel germline mutations in the human E-cadherin (CDH1) gene. Hum Mutat. 2007;28:203. doi: 10.1002/humu.9473. [DOI] [PubMed] [Google Scholar]
  • 29.Risinger JI, Berchuck A, Kohler MF, Boyd J. Mutations of the E-cadherin gene in human gynecologic cancers. Nat Genet. 1994;7:98–102. doi: 10.1038/ng0594-98. [DOI] [PubMed] [Google Scholar]
  • 30.Nass SJ, Herman JG, Gabrielson E, et al. Aberrant methylation of the estrogen receptor and E-cadherin 5′ CpG islands increases with malignant progression in human breast cancer. Cancer Res. 2000;60:4346–4348. [PubMed] [Google Scholar]
  • 31.Droufakou S, Deshmane V, Roylance R, Hanby A, Tomlinson I, Hart IR. Multiple ways of silencing E-cadherin gene expression in lobular carcinoma of the breast. Int J Cancer. 2001;92:404–408. doi: 10.1002/ijc.1208. [DOI] [PubMed] [Google Scholar]
  • 32.Caldeira JR, Prando EC, Quevedo FC, Neto FA, Rainho CA, Rogatto SR. CDH1 promoter hypermethylation and E-cadherin protein expression in infiltrating breast cancer. BMC Cancer. 2006;6:48. doi: 10.1186/1471-2407-6-48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Lipponen P, Saarelainen E, Ji H, Aaltomaa S, Syrjanen K. Expression of E-cadherin (E-CD) as related to other prognostic factors and survival in breast cancer. J Pathol. 1994;174:101–109. doi: 10.1002/path.1711740206. [DOI] [PubMed] [Google Scholar]
  • 34.Charpin C, Garcia S, Bouvier C, et al. E-cadherin quantitative immunocytochemical assays in breast carcinomas. J Pathol. 1997;181:294–300. doi: 10.1002/(SICI)1096-9896(199703)181:3<294::AID-PATH772>3.0.CO;2-V. [DOI] [PubMed] [Google Scholar]
  • 35.Sherr CJ. Cancer cell cycles. Science. 1996;274:1672–1677. doi: 10.1126/science.274.5293.1672. [DOI] [PubMed] [Google Scholar]
  • 36.Tlsty TD, Crawford YG, Holst CR, et al. Genetic and epigenetic changes in mammary epithelial cells may mimic early events in carcinogenesis. J Mammary Gland Biol Neoplasia. 2004;9:263–274. doi: 10.1023/B:JOMG.0000048773.95897.5f. [DOI] [PubMed] [Google Scholar]
  • 37.Holst CR, Nuovo GJ, Esteller M, et al. Methylation of p16(INK4a) promoters occurs in vivo in histologically normal human mammary epithelia. Cancer Res. 2003;63:1596–1601. [PubMed] [Google Scholar]
  • 38.Lewis CM, Cler LR, Bu DW, et al. Promoter hypermethylation in benign breast epithelium in relation to predicted breast cancer risk. Clin Cancer Res. 2005;11:166–172. [PubMed] [Google Scholar]

RESOURCES